You are on page 1of 6

The Journal of Antibiotics (2012) 65, 593–598

& 2012 Japan Antibiotics Research Association All rights reserved 0021-8820/12
www.nature.com/ja

ORIGINAL ARTICLE

Activity of the thiopeptide antibiotic nosiheptide


against contemporary strains of methicillin-resistant
Staphylococcus aureus
Nina M Haste1, Wdee Thienphrapa1, Dan N Tran1, Sandra Loesgen2,5, Peng Sun2, Sang-Jip Nam2,4,
Paul R Jensen2, William Fenical2,3, George Sakoulas1, Victor Nizet1,3 and Mary E Hensler1

The rapid rise in antimicrobial resistance in bacteria has generated an increased demand for the development of novel therapies
to treat contemporary infections, especially those caused by methicillin-resistant Staphylococcus aureus (MRSA). However,
antimicrobial development has been largely abandoned by the pharmaceutical industry. We recently isolated the previously
described thiopeptide antibiotic nosiheptide from a marine actinomycete strain and evaluated its activity against contemporary
clinically relevant bacterial pathogens. Nosiheptide exhibited extremely potent activity against all contemporary MRSA strains
tested including multiple drug-resistant clinical isolates, with MIC values p0.25 mg l 1. Nosiheptide was also highly active
against Enterococcus spp. and the contemporary hypervirulent BI/NAP1/027 strain of Clostridium difficile but was inactive
against most Gram-negative strains tested. Time-kill analysis revealed nosiheptide to be rapidly bactericidal against MRSA in
a concentration- and time-dependent manner, with a nearly 2-log kill noted at 6 h at 10  MIC. Furthermore, nosiheptide was
found to be non-cytotoxic against mammalian cells at 44100  MIC, and its anti-MRSA activity was not inhibited by 20%
human serum. Notably, nosiheptide exhibited a significantly prolonged post-antibiotic effect against both healthcare- and
community-associated MRSA compared with vancomycin. Nosiheptide also demonstrated in vivo activity in a murine model of
MRSA infection, and therefore represents a promising antibiotic for the treatment of serious infections caused by contemporary
strains of MRSA.
The Journal of Antibiotics (2012) 65, 593–598; doi:10.1038/ja.2012.77; published online 10 October 2012

Keywords: contemporary MRSA; marine actinomycete; nosiheptide; thiopeptide

INTRODUCTION multhiomycin, was originally isolated in 1970 and shown to be


Multidrug resistant hospital-associated (HA-) and highly virulent structurally similar to thiostrepton.4,5 Several thiopeptide antibiotics
community-associated (CA-) methicillin-resistant Staphylococcus including nosiheptide block protein synthesis by the inhibition of
aureus (MRSA) have shown increased tolerance or resistance in elongation factors Tu and G.6 Historically, nosiheptide has been used
recent years to vancomycin, linezolid and daptomycin with accom- as a growth-promoting additive in animal feed7 but was never
panying reduction in clinical antimicrobial efficacy. The development developed further as a human therapeutic.
of novel agents without cross-resistance to current antimicrobials Although the discovery of new chemical scaffolds provides the
against MRSA infections is desperately needed.1 potential for novel drugs with new mechanisms of action, the
In our screen of marine-derived actinomycete extract libraries for re-investigation of previously discovered antibacterial scaffolds also
anti-MRSA activity, we identified a potent fraction derived from provides a valuable source of compounds with therapeutic potential.
strain CNT-373, a Streptomycetes species isolated from a marine Surprisingly, despite the structural identification and description of
sediment collected in Fiji. The 1221.16-mol wt active component was Gram-positive antibacterial activity, no detailed characterization of
purified and identified by NMR as the thiopeptide antibiotic nosiheptide activity against contemporary drug-resistant strains such
nosiheptide. Thiopeptide antibiotics encompass a large family of as MRSA has been undertaken. Here, we investigate nosiheptide
compounds that include thiostrepton and nocathiacin and are activity in vitro against a panel of contemporary MRSA and other
comprised of sulfur- and nitrogen-rich heterocycles linked to non- Gram-positive clinical isolates. MRSA killing kinetics and post-
natural amino acids.2,3 Nosiheptide, also referred to historically as antibiotic effects (PAE) are characterized, and nosiheptide in vivo

1Department of Pediatrics, University of California San Diego, La Jolla, CA, USA; 2Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography,
University of California San Diego, La Jolla, CA, USA and 3Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
Correspondence: Dr ME Hensler, Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0687, USA.
E-mail: mhensler@ucsd.edu
4Current address: College of Pharmacy, Suncheon National University, Suncheon 540-950, Republic of Korea.
5Current address: Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA 20892.

Received 6 June 2012; revised 9 August 2012; accepted 15 August 2012; published online 10 October 2012
Nosiheptide activity against contemporary MRSA
NM Haste et al
594

activity is demonstrated. Our results indicate that ‘re-discovered’ Table 1 1H and 13C NMR spectral data of nosiheptide (d in p.p.m.,
natural product antibiotics may harbor meaningful therapeutic J in Hz)
activity against contemporary multidrug resistant pathogens, and Ind CO 180.69 But 3 128.14 6.32 (q, J ¼ 6.84)
therefore warrant further consideration and preclinical development Glu CO 173.52 Pyr 4 127.17 7.64 (s)
in hopes of expanding our current limited pharmacological arsenal. Thz(3) 2 170.18 Thz(5) 5 126.52 8.28 (s)
Thz(4) 2 169.64 Thz(1) 5 125.82 8.47 (s)
MATERIALS AND METHODS Thr CO 169.44 Thz(3) 5 125.67 8.18 (s)
Strain isolation and identification Thz(5) 2 166.83 Ind 6 125.28 7.41 (t, J ¼ 7.57)
Strain CNT-373 was isolated from a marine sediment sample collected at a Thz(2) 2 166.41 Ind 3a 124.36
depth of 5 m off Nacula Island, Fiji. The sediment was dried overnight in a Deala CO 165.99 Thz(2) 5 123.91 7.91 (s)
laminar flow hood and then stamped on agar plates containing medium A1 Thz(1) 2 164.85 Ind 5 122.24 7.12 (d, J ¼ 6.95)
(10 g starch, 4 g yeast extract, 2 g peptone, 16 g agar and 1 l seawater) Thz(3) CO 161.43 Thz(4) 5 120.38 7.66 (s)
supplemented with 100 mg l 1 cyclohexamide to reduce fungal growth. The Thz(2) CO 160.84 Ind 3 118.90
strain was identified as a Streptomyces sp. based on 16S rRNA gene sequence Thz(1) CO 160.57 Ind 7 115.31 7.79 (d, J ¼ 7.04)
analysis (http://eztaxon-e.ezbiocloud.net/ezt_identify) and shares greatest simi- Thz(5) CO 158.59 Deala 3 104.03 6.60 E (d, J ¼ 1.89),
larity (98.3%) with the type strain Streptomyces althioticus. This low level of 5.58 Z (d, J ¼ 1.89)
sequence identity suggests it may be a new species. The 16S rRNA sequence has
Thz(4) 4 155.00 Thr 3 67.45 4.04 (m)
been deposited in GenBank under accession number JQ946086.
Pyr 3 150.30 Glu 4 67.18 4.14 (d, J ¼ 11.54)
Thz(1) 4 149.81 Ind 4’ 66.60 5.85 (d, J ¼ 11.25),
Fermentation and extraction 5.02 (d, br, J ¼ 10.79)
A 2-ml-frozen glycerol stock of strain CNT-373 was used to inoculate 25 ml of Thz(5) 4 149.76 Thr 2 56.75 4.36 (d, J ¼ 7.79)
liquid A1 medium and shaken at 230 r.p.m. and 27 1C. After 5 days, the seed Thz(3) 4 148.45 Cys 2 50.17 5.98 (d, br, J ¼ 9.00)
culture was used to inoculate a 1-l culture in growth medium A1bfe þ C (A1
Thz(2) 4 147.86 Glu 2 45.85 5.76 (t, J ¼ 10.19)
medium with the addition of 1 g CaCO3, 100 mg KBr and 40 mg Fe2(S-
Pyr 6 143.99 Glu 3 36.72 2.35 S (m), 1.80 R (m)
O4)34H2O). After 3 days of shaking, 25-ml aliquots were used to inoculate 18
Ind 7a 137.44 Cys 3 29.71 3.79 (dd, J ¼ 3.15, 12.06),
Fernbach flasks (2.8 l) each containing 1 l medium A1bfe þ C. After 7 days of
3.71 (dd, J ¼ 4.50, 12.00)
cultivation, sterilized XAD-16 resin (20 g l 1) was added, shaken for 6 h and
Pyr 2 134.46 Thr CH3 17.52 0.95 (s)
collected by filtration through cheesecloth. The resin was then washed with
deionized water and eluted with acetone. The acetone solvent was removed Deala 2 132.94 But CH3 14.58 1.68 (d, J ¼ 7.00)
under reduced pressure and the resulting aqueous layer extracted with ethyl Ind 2 130.64 Ind CH3 11.96 2.54 (s)
acetate (3  500 ml). The combined ethyl acetate extracts were dried over Pyr 5 130.44 Ind NH 10.83 (s, br)
anhydrous sodium sulfate, decanted and concentrated to dryness to yield 2.4 g But 2 129.41 Deala NH 9.99 (s)
of crude material. Ind 4 128.73 But NH 9.76 (s)
Glu NH 7.97 (d, br, J ¼ 7.29)
Isolation of nosiheptide Glu OH not observed
The crude extract was dissolved in methanol and dichloromethane, adsorbed Cys NH 7.82 (s)
onto silica gel (2.5 g), and fractionated on a short column of silica gel Thr NH 7.50 (s)
(6  2.5 cm, h  w) using a 10% step gradient from 100% isooctane to 100% Thr OH 7.48 (s)
ethyl acetate as eluents. Fractions with antibacterial activity were eluted with 90
and 100% ethyl acetate. The active fractions were pooled and subjected to a
size exclusion column chromatography with Sephadex LH-20 using methanol
as eluent. The final purification step was achieved with silicia gel, using ethyl
acetate with 10% dimethylformamide to give nosiheptide (210 mg).

Spectroscopic analysis of nosiheptide


1H, 13C and 2D NMR spectroscopic data were obtained on a Varian Inova 500-

MHz spectrometer in a solvent mixture of methanol-d4 and CDCl3 (3:1) to


facilitate solubility. Offline processing was conducted using topspin NMR
software by Bruker BioSpin 2011 (iNMR, http://www.inmr.net). The NMR
data (Table 1) are in good agreement with previously published NMR data for
nosiheptide,8 with the resultant nosiheptide structure shown (Figure 1). High-
resolution ESI-TOF mass spectra were provided by the MS facility at the
Department of Chemistry and Biochemistry at the University of California
San Diego, CA: HR-ESI-TOF-MS [M þ H] þ m/z 1222.1565 (calcd for
C51H44N13O12S6 1222.1551, D1.13 p.p.m.). Low resolution LC-MS data were
measured using a Hewlett-Packard HP1100 integrated LC/MS system with
a reversed-phase C18 column (Phenomenex Luna, 4.6 mm  100 mm, 5 mm, Figure 1 Structure of marine-derived nosiheptide.8 A full color version of
Phenomenex, Torrance, CA, USA) at a flow rate of 0.7 ml min 1. this figure is available at The Journal of Antibiotics journal online.

Bacterial strains and susceptibility testing


Details on the strains used in this study are in Table 2. MRSA Sanger 252 USA according to Clinical and Laboratory Standards Institute methods.9 The MIC
200 strain was obtained through the Network of Antimicrobial Resistance in for Clostridium difficile was determined by the agar dilution reference method
S. aureus (NARSA) program supported under NIAID/NIH contract no. according to CLSI guidelines.10,11 Susceptibility of nosiheptide against MRSA
HHSN272200700055C. Susceptibility testing was performed in duplicate using strain TCH1516 was also determined in the presence of 20% activated-pooled
cation-adjusted Mueller–Hinton broth (CA-MHB) and Mueller–Hinton agar human serum (freshly collected from consenting healthy donors) and 80% MHB

The Journal of Antibiotics


Nosiheptide activity against contemporary MRSA
NM Haste et al
595

Table 2 Nosiheptide MIC

Strain Details MIC (mg l 1)

TCH1516 USA 300 CA-MRSA (ATCC) 0.06 (0.06)a


Sanger 252 USA 200 HA-MRSA (NARSA) 0.03
ATCC 33591 HA-MRSA 0.06
A593720 Bloodstream MRSA, progenitor to A5940 0.125
A5940 VISA20 In vivo VISA 0.125
A630020 MRSA, progenitor to A6298 0.125
A6298 VISA20 VISA 0.125
SA85312 Progenitor to SI853b 0.125
SA853b12 In vitro VISA 0.125
HIP5836 NJ VISA12,20 VISA—NJ 0.125
VRSA PA12 vanA vancomycin-resistant Staphylococcus aureus 0.06
VRSA MI12 vanA vancomycin-resistant S. aureus 0.125
A781712 Progenitor to A7819, A7819erm 0.06
A7819LinR 12 LinezolidR isolate 0.06
A7819ermR 12 Erythromycin-resistant variant (plasmid) 0.06
SA35412 Progenitor to SA355 0.125
SA35512 LinezolidR isolate 0.125
061612 (DapS) Endocarditis MSSA, progenitor to 0701 0.125
070112 (DapR) Daptomycin-resistant MSSA 0.06
MSSA ATCC 29213 0.125
RN912020 MSSA, Agr knockout, progenitor to RN9120b 0.06
RN9120V20 VISA selected in vitro 0.25
Staphylococcus epidermidis ATCC 12228 0.5
VRE-CUS VRE bloodstream left-sided endocarditis 0.125
VRE-WMC VRE bloodstream infection BI/NAP1/027 Hypervirulent Contemporary 0.125
Clostridium difficile BI Clostridium difficile 0.008
Moraxella catarrhalis ATCC 25238 0.5
Eseudomonas coli 1035 21 Clinical respiratory tract isolate 44
Pseudomonas aeruginosa ATCC 27853 44
Acinetobacter baumannii ATCC 19606 44
Enterobacter cloacae ATCC 13047 44

Abbreviations: ATCC, American Type Culture Collection; VRE-CUS, vancomycin-resistant Enterococcus faecium.
aMIC in 20% human serum.

by adding resazurin and assessing the conversion to resorufin exactly as previ- Mammalian cell cytotoxicity
ously described.12 This method provides a visual readout of color change from Mammalian cell cytotoxicity was assessed as previously described.13 Briefly,
the blue indicator resazurin to the pink resorufin, a sign of bacterial growth. 2  104 HeLa cells (American Type Culture Collection no. CCL-2, ATCC,
Manassas, VA, USA) were seeded per well of sterile 96-well tissue culture-
treated plates (Falcon; Becton Dickinson, Franklin Lakes, NJ, USA). After 24 h,
Time-kill studies
the medium (RPMI containing 10% heat-inactivated fetal bovine serum) was
Time-kill studies were done essentially as previously described in this
replaced with fresh medium containing increasing concentrations of
laboratory.13 Briefly, nosiheptide was added at multiples of the MIC in CA-
nosiheptide up to 128 mg l 1, and the plates were incubated at 37 1C in 5%
MHB. MRSA strains or vancomycin-resistant Enterococcus faecium were added
CO2. Cell viability was assayed at 72 h by measuring the reduction of MTS
at a starting inoculum of 5  105 cfu ml 1, and the cultures were incubated in
(3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-
a 37 1C shaking incubator (New Brunswick Scientific, Enfield, CT, USA).
2H-tetrazolium) using the CellTiter 96 Aqueous nonradioactive cell
Samples were taken at specified timepoints for enumeration of viable bacteria
proliferation assay according to the manufacturer’s instructions (Promega,
by plating serial dilutions on Todd–Hewitt agar plates.
Madison, WI, USA).

Post-antibiotic effect
PAE was assessed using the viable plate count method as previously In vivo testing
described.13 For these studies, either nosiheptide or vancomycin was added A murine model of i.p. infection was used to test in vivo efficacy of
at 10  their respective MICs to 14 ml Falcon tubes containing CA-MHB. CA- nosiheptide.15 Eight-week-old female CD1 mice (Charles River, Wilmington,
MRSA strain TCH1516 or HA-MRSA strain Sanger 252 was added at 5  105 MA, USA) were injected i.p. with 1–2  109 cfu of the HA-MRSA strain Sanger
cfu ml, and the tubes were incubated under shaking conditions at 37 1C for 1 h. 252 in 4% hog gastric mucin. The mice were treated with nosiheptide
The bacteria were then pelleted and washed in 4 ml of antibiotic-free CA- (20 mg kg 1) i.p. at 1 and 8 h after bacterial inoculation and were monitored
MHB. The pellets were resuspended in 4 ml CA-MHB and incubated at 37 1C for mortality and clinical status twice daily thereafter for 5 days. Moribund mice
in a shaking incubator. Surviving bacteria were assessed at specified timepoints were humanely killed as were mice at the end of the study. These experiments
as for the time-kill assay. The PAE was determined as previously described.13,14 were reviewed and approved by the Animal Subjects Committee of UCSD.

The Journal of Antibiotics


Nosiheptide activity against contemporary MRSA
NM Haste et al
596

Statistical analysis favorably with vancomycin kinetics at 10  and 20  their respective


The data from the survival studies were analyzed with GraphPad Prism MICs (Figure 2d).
(GraphPad Software, Incorporated, La Jolla, CA, USA) using log-rank analysis
(Mantel–Cox test). Data were considered significant at Po0.05. Prolonged PAE and lack of cytotoxicity
Given its favorable MRSA killing kinetics, nosiheptide was assessed
RESULTS for its PAE. Notably, nosiheptide exhibited prolonged PAEs against
In vitro antimicrobial activity both HA- and CA-MRSA compared with the most commonly used
Nosiheptide exhibited strong activity against all contemporary MRSA systemic antibiotic, vancomycin, at 10  their respective MICs
and MSSA strains tested including multidrug-resistant clinical isolates (Figures 3a and b). The PAE for nosiheptide was calculated to exceed
(Table 2), with MIC values p0.25 mg l 1. Notably, clinical MRSA 9 h for both HA- and CA-MRSA. We also tested nosiheptide for
isolates previously demonstrated to have developed resistance to in vitro evidence of mammalian cell cytotoxicity as a prelude to our
front-line antibiotics including daptomycin, linezolid and vancomycin in vivo experiments. No evidence for nosiheptide cytotoxicity was
remained highly sensitive to nosiheptide. This activity against MRSA observed when incubated for 72 h with the cervical carcinoma HeLa
was also completely unaffected by the presence of 20% human serum cell line at up to 128 mg l 1, which is B1000-fold above the MIC
(Table 2). Nosiheptide was also highly active against Enterococcus spp. against MRSA. We also found that nosiheptide activity against
and the Gram-negative pathogen Moraxella catarrhalis, but did not USA300 MRSA was not inhibited in 20% human serum (Table 2).
possess activity at p4 mg l 1 against the other Gram-negative strains
tested including clinical isolates of Escherichia coli and Pseudomonas In vivo activity
aeruginosa. Addressing another critical antibiotic resistance challenge, Owing to its potent anti-MRSA activities in vitro, nosiheptide was
nosiheptide showed extremely potent activity against the contempor- tested in a murine model of i.p. MRSA infection. Mice were infected
ary hypervirulent BI strain of C. difficile (also known as the NAP1 or with HA-MRSA strain Sanger 252, followed by nosiheptide treatment
ribotype 027 strain). (20 mg kg 1, i.p.) at 1 and 8 h post infection. Although all mice
became lethargic and exhibited piloerection within B4 h of infection,
Time-kill kinetics nosiheptide provided significant (Po0.03) protection against mor-
In vitro time-kill analysis was used to assess nosiheptide killing tality (Figure 4). Ten out of 10 of the nosiheptide-treated mice
kinetics. Nosiheptide was rapidly bactericidal against MRSA in a remained alive on day 3, whereas 6/10 of the controls died on day 1.
concentration- and time-dependent manner (Figure 2a), with a nearly By the end of the study, only one mouse in the nosiheptide group had
2-log kill noted within 6 h at both 10  and 20  MIC. A 2-log kill of died. These results provide evidence of significant in vivo activity for
MRSA was also noted for nosiheptide against MRSA resistant to other nosiheptide.
antibiotics, such as linezolid or erythromycin (Figure 2b). Although
nosiheptide demonstrated bactericidal activity against MRSA at 10  DISCUSSION
and 20  the MIC, it was bacteriostatic against a clinical bloodstream In our search for new anti-MRSA antibiotics from marine-derived
isolate of vancomycin-resistant Enterococcus faecium up to 64  MIC microorganisms, we identified an actinomycete strain that produced a
(Figure 2c). The MRSA killing kinetics of nosiheptide also compared metabolite shown to be nosiheptide. Although the structure of this

10 10
None A7817
8 Nosiheptide 20X MIC 8 A7819
log10 cfu/ml

log10 cfu/ml

Nosiheptide 10X MIC A7819em


6 Nosiheptide 1X MIC 6 SI354
SI354-LinR
4 4 ATCC33591
2 2

0 0
0 3 6 9 12 15 18 21 24 0 3 6 9 12 15 18 21 24
Time (hr) Time (hr)

10 10
None None
8 Nosiheptide 2X MIC 8 Nosiheptide 20X MIC
log10 cfu/ml

log10 cfu/ml

Nosiheptide 4X MIC Vancomycin 20X MIC


6 6 Nosiheptide 10X MIC
Nosiheptide 8X MIC
Vancomycin 10X MIC
4 Nosiheptide 32X MIC 4
Nosiheptide 64X MIC
2 2

0 0
0 3 6 9 12 15 18 21 24 0 3 6 9 12 15 18 21 24
Time (hr) Time (hr)

Figure 2 Time-kill kinetics of nosiheptide against MRSA and VRE. Nosiheptide killing kinetics was studied against MRSA and VRE at multiples of their
MICs (MIC ¼ 0.06 mg l 1 for nosiheptide). (a) Killing kinetics of nosiheptide at 1  , 10  and 20X MIC against MRSA USA300 strain TCH1516
(MIC ¼ 0.06 mg l 1). (b) Nosiheptide killing kinetics against various multi-drug resistant MRSA at 10  their respective MICs. (c) Nosiheptide activity over
time against vancomycin-resistant Enterococcus faecium at multiples of the MIC (MIC ¼ 0.125 mg l 1). (d) Nosiheptide time-kill kinetics against MRSA
USA300 strain TCH1516 compared with vancomycin at 10  and 20  their respective MICs (MIC ¼ 0.06 mg l 1 for nosiheptide; MIC ¼ 1.56 mg l 1 for
vancomycin). Each assay was repeated in duplicate (for VRE) or triplicate (for MRSA), and the data shown are the mean±s.d. from one representative
experiment.

The Journal of Antibiotics


Nosiheptide activity against contemporary MRSA
NM Haste et al
597

CA-MRSA contemporary MRSA strains tested regardless of their resistance


to other front-line therapeutics including vancomycin, linezolid and
10 Nosiheptide 10X MIC daptomycin. Importantly, high-level activity was also noted against
8 Vancomycin 10X MIC the increasingly problematic pathogen, C. difficile, especially signifi-
log10 cfu/ml

None
cant as the FDA has approved only one new drug (Fidaxomicin) to
6
treat C. difficile in the past two decades. Although the activity of
4 some thiazole antibiotics may be reduced in the presence of serum,
2
we found that nosiheptide activity against USA300 MRSA was
unaffected in the presence of 20% human serum, suggesting that
0 serum inhibition may not necessarily be a characteristic of all
0 3 6 9 12 15 18 21 24 thiazole-containing antibiotics.
Time (hr) Nosiheptide exhibited favorable MRSA killing kinetics, a very
prolonged PAE, and was active in a murine model of i.p. infection.
HA-MRSA
Although in vivo evidence of nosiheptide activity has not been
10 published, Benazet et al.16 report that nosiheptide protected mice
Nosiheptide 10X MIC
8 Vancomycin 10X MIC from S. aureus mortality only when the compound was administered
at the site of infection. However, no data were presented to support
log10 cfu/ml

None
6 this claim. We report in the current paper nosiheptide activity when
4 the compound is administered 1 and 8 h after MRSA injection at the
site of infection as well (Figure 4). Our data are consistent with the
2 claims of Benazet et al.16 and point to the fact that nosiheptide is
0 efficacious in vivo but may either be metabolized or not distributed
0 3 6 9 12 15 18 21 24 (due to localized precipitation or lack of absorption) when injected. A
Time (hr) closely related compound, glycothiohexide a, was once shown to be
quite active against MRSA and vancomycin-resistant E. faecium.17–19
Figure 3 PAE of nosiheptide on contemporary MRSA strains. CA-MRSA
strain TCH-1516 (a) or HA-MRSA strain Sanger 252 (b) were incubated for
However, despite its in vitro potency, this compound had minimal
1 h with nosiheptide or vancomycin at 10  MIC. The antibiotics were activity in a S. aureus model of i.p. infection when administered s.c.
removed, and the bacteria were washed and allowed to recover in antibiotic- It is unknown whether in the case of glycothiohexide a, pharmaco-
free media. The curves show the rates of growth following antibiotic kinetic and/or possible metabolic factors abrogated potential efficacy.
treatment. The data represent the mean±s.d. of one representative assay In pilot studies, we also found that nosiheptide exhibited significantly
repeated in duplicate. reduced activity when injected s.c. (data not shown) following i.p.
MRSA inoculation and are currently embarking upon collaborative
studies of liposomal and other lipophilic drug formulations to
100 Nosiheptide optimize nosiheptide delivery in vivo.
Control In sum, nosiheptide represents a promising antibiotic scaffold for
80 further development owing to its potent anti-MRSA and other Gram-
% Survival

60
positive activity, lack of inhibition by human serum, lack of
mammalian cell cytotoxicity and demonstrated tolerability when
40 delivered orally to animals.7 Many of the synthesis/manufacturing
issues have already been addressed, as nosiheptide is produced in
20 mass quantities by manufacturers worldwide. Cumulatively, these
0 properties provide nosiheptide an advantage over other early-stage
0 1 2 3 4 5 antibacterials, many of which are later found to be cytotoxic or
Day serum-inhibited, or are fraught with difficulties in scale-up of
production.
Figure 4 Nosiheptide protection in a murine model of i.p. MRSA infection.
Plot showing survival of mice (n ¼ 10 per group) infected i.p. with HA- ACKNOWLEDGEMENTS
MRSA strain Sanger 252, followed by i.p. treatment with either nosiheptide
NMH was supported by the National Institutes of Health (NIH) Training
(20 mg kg 1, filled circles) or vehicle control (filled squares) at 1 h or 8 h
Program in Marine Biotechnology (T32 GM067550) and a Ruth L Kirschstein
after infection (represented by upward arrows on the x axis). N ¼ 10 mice
National Research Service Award (NRSA) from National Institutes of Health
per treatment group (Po0.03 by log-rank analysis).
grants (5 F31 GM090658-02). SL was supported by the German Research
Foundation (DFG). WF, VN, PRJ and MEH were supported by National
Institutes of Health grant GM084350. PRJ and WF acknowledge financial
compound was originally elucidated several decades ago and activity support from the NIH Fogerty Center International Cooperative Biodiversity
against Gram-positive activity was noted, a paucity of data exist on Groups program (grant U01-TW00007-401). We thank K Freel and C
Kauffman for assistance with fieldwork and W Aalbersberg (University of the
the therapeutic potential for this compound given contemporary
South Pacific) for providing laboratory space and facilitating the field
concerns of multi-drug resistant bacterial pathogens. With growing collections. We gratefully acknowledge the people of Fiji for their hospitality
antibiotic resistance exhibited by MRSA and other Gram-positive and permission to collect samples from their local waters. We are also grateful
pathogens driving the need for new antimicrobials, we sought to Diane M Citron and Ellie Goldstein MD of RM Alden Research
to further characterize this compound for its activity against Laboratories (Culver City, CA, USA) for performing susceptibility testing
HA- and CA-MRSA. Nosiheptide was exquisitely active against all against Clostridium difficile.

The Journal of Antibiotics


Nosiheptide activity against contemporary MRSA
NM Haste et al
598

11 Goldstein, E. J. C. et al. Comparative susceptibilities to fidaxomicin (OPT-80) of


1 Dhand, A. et al. Use of antistaphylococcal beta-lactams to increase daptomycin activity isolates collected at baseline, recurrence, and failure from patients in two Phase III
in eradicating persistent bacteremia due to methicillin-resistant Staphylococcus trials of fidaxomicin against Clostridium difficile infection. Antimicrob. Agents Che-
aureus: role of enhanced daptomycin binding. Clin. Infect. Dis. 53, 158–163 mother. 55, 5194–5199 (2011).
(2011). 12 Sakoulas, G. et al. Novel bacterial metabolite merochlorin A demonstrates in vitro
2 Bagley, M. C., Dale, J. W., Merritt, E. A. & Xiong, X. Thiopeptide antibiotics. Chem. Rev. activity against multi-drug resistant methicillin resistant Staphylococcus aureus. PloS
105, 685–714 (2005). One 7, e29439 (2012).
3 Nicolaou, K. C., Chen, J. S., Edmonds, D. J. & Estrada, A. A. Recent advances in the 13 Haste, N. M. et al. Pharmacological properties of the marine natural product
chemistry and biology of naturally occurring antibiotics. Angew. Chem. Int. Ed. Engl. marinopyrrole A against methicillin resistant Staphylococcus aureus. Antimicrob.
48, 660–719 (2009). Agents Chemother. 55, 3305–3312 (2011).
4 Tanaka, T., Endo, T., Shimazu, A., Yoshida, R. & Suzuki, Y. A new antibiotic, 14 Craig, W. A. & Gundmundsson, S. in Antibiotics in Laboratory Medicine (ed. Lorian, V.)
multhiomycin. J. Antibiot. 23, 231–237 (1970). 4th edn. 296–329 (Williams and Wilkins, Baltimore, MD, 1996).
5 Tanaka, T., Sakaguchi, K. & Yonehara, H. On the mode of action of multhiomycin, I. 15 Haste, N. M. et al. Activity of the streptogramin antibiotic etamycin against methicillin-
Effects of multhiomycin on macromolecular syntheses. J. Antibiot. 23, 401–407 resistant Staphylococcus aureus. J. Antibiot. 63, 219–224 (2010).
(1970). 16 Benazet, F. et al. Nosiheptide, a sulfur-containing peptide antibiotic isolated from
6 Cundliffe, E. & Thompson, J. The mode of action of nosiheptide (multhiomycin) and Streptomyces actuosus 40037. Experientia 36, 414–416 (1980).
the mechanism of resistance in the producing organism. J. Gen. Microbiol. 126, 17 Northcote, P. T. et al. Glycothiohexide alpha, a novel antibiotic produced by ‘Sebekia’
185–192 (1981). sp., LL-14E605. II. Isolation and physical-chemical characterization. J. Antibiot. 47,
7 Benazet, F. & Cartier, J. R. Effect of nosiheptide as a feed additive in chicks on the 894–900 (1994).
quantity, duration, prevalence of excretion, and resistance to antibacterial agents of 18 Northcote, P. T., Siegel, M., Borders, D.B. & Lee, M.D. Glycothiohexide alpha, a novel
Salmonella typhimurium; on the proportion of Escherichia coli and other coliforms antibiotic produced by ‘Sebekia’ sp., LL-14E605. III. Structural elucidation.
resistant to antibacterial agents; and on their degree of resistance. Poult. Sci. 59, J. Antibiot. 47, 901–908 (1994).
1405–1415 (1980). 19 Steinberg, D. A. et al. Glycothiohexides, novel antibiotics produced by ‘Sebekia’ sp. LL-
8 Mocek, U. et al. 1H and 13C NMR Assignments of the thiopeptide antibiotic 14E605. I. Taxonomy, fermentation and biological evaluation. J. Antibiot. 47,
nosiheptide. J. Antibiot. 42, 1643–1648 (1989). 887–893 (1994).
9 Clinical and Laboratory Standards Institute. Methods for Dilution Antimicrobial 20 Sakoulas, G. et al. Accessory gene regulator (agr) locus in geographically diverse
Susceptibility Tests for Bacteria that Grow Aerobically. Document M7–A7 (Clinical Staphylococcus aureus with reduced susceptibility in vancomycin. Antimicrob. Agents
and Laboratory Standards Institute, Wayne, PA, 2006). Chemother. 46, 1492–1502 (2002).
10 Clinical and Laboratory Standards Institute. Methods for Antimicrobial Susceptibility 21 Laplante, K. L. & Sakoulas, G. Evaluating aztreonam and ceftazidime pharmacodynamics
Testing of Anaerobic Bacteria. Approved Standard M11-A7 (Clinical and Laboratory with Escherichia coli in combination with daptomycin, linezolid, or vancomycin in an
Standards Institute, Wayne, PA, 2007). in vitro pharmacodynamic model. Antimicrob. Agents Chemother. 53, 4549–4555 (2009).

The Journal of Antibiotics

You might also like